CN112625025A - Pyridyl substituted quinoline derivative and its prepn and use - Google Patents

Pyridyl substituted quinoline derivative and its prepn and use Download PDF

Info

Publication number
CN112625025A
CN112625025A CN202011640042.3A CN202011640042A CN112625025A CN 112625025 A CN112625025 A CN 112625025A CN 202011640042 A CN202011640042 A CN 202011640042A CN 112625025 A CN112625025 A CN 112625025A
Authority
CN
China
Prior art keywords
compound
substituted
formula
radical
membered
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202011640042.3A
Other languages
Chinese (zh)
Other versions
CN112625025B (en
Inventor
邹敏
金波
郑立运
王明升
李佳雯
王强
方先珍
张壮丽
夏书梼
黄小婉
郭桐
张晓�
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Henan Academy of Medical and Pharmaceutical Sciences
First Affiliated Hospital of Zhengzhou University
Original Assignee
Henan Academy of Medical and Pharmaceutical Sciences
First Affiliated Hospital of Zhengzhou University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Henan Academy of Medical and Pharmaceutical Sciences, First Affiliated Hospital of Zhengzhou University filed Critical Henan Academy of Medical and Pharmaceutical Sciences
Priority to CN202011640042.3A priority Critical patent/CN112625025B/en
Publication of CN112625025A publication Critical patent/CN112625025A/en
Application granted granted Critical
Publication of CN112625025B publication Critical patent/CN112625025B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The invention belongs to the field of anti-cancer drugs, and particularly relates to a pyridyl substituted quinoline derivative, and a preparation method and application thereof. The present invention provides compounds of formula I, stereoisomers or pharmaceutically acceptable salts thereof:

Description

Pyridyl substituted quinoline derivative and its prepn and use
Technical Field
The invention belongs to the field of anti-cancer drugs, and particularly relates to a pyridyl substituted quinoline derivative, and a preparation method and application thereof.
Background
Most cancers are fatal diseases with high mortality, and the incidence of these diseases is currently on the rise in all ages, particularly in developed countries. The world health organization indicates that cancer is one of the leading causes of human death, and the worldwide annual number of cancer deaths is expected to increase to 1310 ten thousand by 2030.
Protein tyrosine kinases are capable of catalyzing the phosphorylation of specific tyrosyl residues in a variety of proteins involved in the regulation of cell growth and differentiation. Protein tyrosine kinases can be broadly classified as receptor (e.g., EGFR, HER2(C-ERB-2), C-MET, TIE-2, PDGFR, FGFR) or non-receptor (e.g., C-SRC, LCK, ZAP70) kinases. Inappropriate or uncontrolled activation of many of these kinases (e.g., aberrant protein tyrosine kinase activity resulting from overexpression or mutation) has been shown to result in uncontrolled cell growth. Abnormal activity of protein tyrosine kinases such as HER2, C-SRC, C-MET, EGFR, PDGFR and the like is associated with human malignancies. For example, increased EGFR activity is associated with non-small cell lung cancer, bladder cancer, and head and neck cancer, and increased HER2 activity is associated with cancers of the breast, ovary, stomach, and pancreas. Thus, inhibition of protein tyrosine kinases should provide a treatment for the above-mentioned tumors.
At present, the defects of nonselectivity, acute toxicity or high cell drug resistance of the antitumor drugs generally exist. Research shows that the multi-target anticancer drug is expected to improve the treatment effect of single-target anticancer drugs and reduce drug resistance, so that the multi-target anticancer drug is an important research direction for research and development of anticancer drugs. Therefore, there is a need to develop new anti-cancer drugs with multiple targets to enrich the choice of treatment options and/or to improve the problems of existing anti-cancer drugs.
Disclosure of Invention
To ameliorate the above problems of the prior art, the present invention provides a compound represented by the following formula I:
Figure BDA0002878186340000021
wherein R is1Selected from CN, halogen, OH, SH, unsubstituted or substituted by one or more RaSubstituted of the following groups: c1-40Alkyl radical, C1-40Alkyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each Y is the same or different and is independently selected from a bond or-O-;
R2、R3identical or different, independently of one another, from H, unsubstituted or substituted by one or more RbSubstituted of the following groups: c1-40Alkyl radical, C3-40Cycloalkyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl;
R4selected from unsubstituted or substituted by one or more RcSubstituted of the following groups: 5-20 membered heteroaryl or 3-20 membered heterocyclyl, with the proviso that said heteroaryl isOr at least 1 ring-constituting atom of the heterocyclic group is N;
R5selected from unsubstituted or substituted by one or more RdSubstituted C1-40An alkyl group;
each Ra、Rb、Rc、RdIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2Unsubstituted or substituted by one or more ReSubstituted of the following groups: c1-40Alkyl radical, C2-40Alkenyl radical, C3-40Cycloalkyl radical, C3-40Cycloalkenyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, C1-40Alkyloxy, C2-40Alkenyloxy radical, C3-40Cycloalkyl oxy, C3-40Cycloalkenyloxy, C6-20Aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each ReIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2Unsubstituted or substituted by one or more RfSubstituted of the following groups: c1-40Alkyl radical, C2-40Alkenyl radical, C3-40Cycloalkyl radical, C3-40Cycloalkenyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, C1-40Alkyloxy, C2-40Alkenyloxy radical, C3-40Cycloalkyl oxy, C3-40Cycloalkenyloxy, C6-20Aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each RfIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2、C1-40Alkyl radical, C2-40Alkenyl radical, C3-40Cycloalkyl radical, C3-40Cycloalkenyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, C1-40Alkyloxy, C2-40Alkenyloxy radical, C3-40Cycloalkyl oxy, C3-40Cycloalkenyloxy, C6-20Aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each R6、R7Same or different, independently from each other selected from H, C1-40Alkyl radical, C3-40Cycloalkyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl.
According to an embodiment of the invention, in formula I
Figure BDA0002878186340000032
Represents R4The other groups on the double bond to which they are bonded may be in either the cis (Z) or trans (E) configuration.
For example, the compound of formula I may have the structure shown in formula I':
Figure BDA0002878186340000031
wherein R is1、R2、R3、R4、R5Having the definitions as described above.
According to an embodiment of the invention, R1Selected from CN, halogen, OH, SH, C1-6Alkyl, NH2、COOH、-OC(O)C1-6Alkyl, -C (O) OC1-6An alkyl group.
According to an embodiment of the invention, R2、R3Identical or different, independently of one another, from H or by one or more RbSubstituted C6-10And (4) an aryl group.
According to an embodiment of the invention, RbSelected from halogen, CN, OH, SH, NO2Unsubstituted or substituted by one or more ReSubstituted of the following groups: c1-6Alkyl radical, C1-6An alkyloxy group.
According to the inventionEmbodiment (I) ReSelected from halogen, CN, OH, SH, NO2Unsubstituted or substituted by one or more RfSubstituted C6-10And (4) an aryl group.
According to an embodiment of the invention, RfSelected from halogens.
According to an embodiment of the invention, the halogen is selected from F, Cl, Br or I.
According to an exemplary embodiment of the invention, R2Selected from 4- (benzyloxy) phenyl, 4-chlorophenyl, 4- (3-fluorobenzyloxy) -3-chlorophenyl.
According to an embodiment of the invention, R4Selected from unsubstituted or substituted by one or more RcA substituted 5-6 membered heteroaryl or 5-6 membered heterocyclyl, provided that 1 or 2 ring atoms of the heteroaryl or heterocyclyl are N. As an example, R4Selected from unsubstituted or substituted by one or more RcSubstituted of the following groups: pyridin-1-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl.
According to an embodiment of the invention, R5Is selected from C1-6An alkyl group.
According to an embodiment of the invention, each R isa、Rb、Rc、RdIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2、C1-6Alkyl radical, C1-6Alkyloxy, C6-10Aryloxy, 5-6 membered heteroaryloxy, 5-6 membered heterocyclyloxy.
According to a preferred embodiment of the invention, the compound of formula I has the structure shown in formula I-1 below:
Figure BDA0002878186340000041
Figure BDA0002878186340000051
according to a more preferred embodiment of the invention, the compound of formula I has the structure shown in formula I-2 below:
Figure BDA0002878186340000052
according to a more preferred embodiment of the invention, the compound of formula I is selected from the following compounds:
(E) -N- (4- (4- (benzyloxy) anilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide (a);
(E) -N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide (b);
(E) -N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide (c);
(E) -N- (4- (4-fluoroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide (d).
The invention also provides a preparation method of the compound shown in the formula I, which comprises the step of reacting the compound shown in the formula II with the compound shown in the formula III to obtain the compound shown in the formula I:
Figure BDA0002878186340000053
wherein R is1、R2、R3、R4And R5Having the definitions as described above.
According to an embodiment of the present invention, the reaction of the compound of formula II with the compound of formula III may be carried out in the presence of an organic solvent, which may be selected from N-methylpyrrolidone.
According to an embodiment of the present invention, the reaction temperature of the compound of formula II and the compound of formula III may be 0 to 10 ℃, and the reaction time may be 1 to 5 hours; the product of the reaction may be purified by recrystallization, which may be a mixed solvent of dichloromethane and methanol, wherein the volume ratio of dichloromethane to methanol may be 1:1 to 1: 5.
According to an embodiment of the invention, the compound of formula II is prepared by reaction of a compound of formula IV below with a halogenating agent:
Figure BDA0002878186340000061
wherein R is4Having the definitions as described above.
According to an embodiment of the present invention, the halogenating agent may be selected from, for example, one of a phosphorus halide, a sulfoxide halide (e.g., thionyl chloride), a triphenylphosphine halide, an oxalyl halide; the reaction temperature of the compound of formula IV with the halogenating agent can be in the range of 10-78 deg.C; the reaction time can be 1-10 h; the reaction may be carried out in the presence or absence of an additional organic solvent, and preferably, the halogenating agent may serve as a solvent for the above reaction.
According to an embodiment of the invention, the compound of formula III is prepared by the reaction of a compound of formula V below with a compound of formula VI below:
Figure BDA0002878186340000062
wherein L is selected from a leaving group, such as Cl, Br or I;
R1、R2、R3、R4and R5Having the definitions as described above.
According to an embodiment of the present invention, the reaction temperature of the compound of formula V with the compound of formula VI may be 10 to 90 ℃ and the reaction time may be 10 to 20 hours.
According to an embodiment of the present invention, the reaction of the compound of formula V with the compound of formula VI may be carried out in the presence of an organic solvent, which may be selected from alcohol compounds, for example one or more selected from isopropanol, methanol or ethanol.
According to a preferred embodiment of the present invention, the reaction product of the compound of formula V and the compound of formula VI may be purified by recrystallization in an organic solvent, which may be selected from alcoholic compounds, for example from one or more of isopropanol, methanol or ethanol.
According to an embodiment of the present invention, a step of forming an acid addition salt of the compound represented by formula I with an acid may also be included.
The invention also provides a pharmaceutical composition, which comprises a therapeutically effective amount of the compound shown in the formula I, a stereoisomer or a pharmaceutically acceptable salt thereof.
According to the present invention, the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers or excipients.
According to the present invention, the pharmaceutical composition may further comprise one or more additional therapeutic agents.
Also provided herein is a method of inhibiting cell proliferation in vitro or in vivo, comprising contacting a cell with an effective amount of a compound of formula I, a stereoisomer, or a pharmaceutically acceptable salt thereof, as defined herein.
The invention also provides a method for preventing or treating EGFR and/or HER2 kinase mediated diseases, which comprises administering to a patient a prophylactically or therapeutically effective amount of a compound as shown in formula I, a stereoisomer, or a pharmaceutically acceptable salt thereof.
Also provided herein is a method of treating cancer and/or inhibiting metastasis associated with cancer in a patient in need of such treatment, comprising administering to the patient a prophylactically or therapeutically effective amount of a compound of formula I, a stereoisomer, or a pharmaceutically acceptable salt thereof, as defined herein.
The invention also provides application of the compound shown in the formula I, the stereoisomer or the pharmaceutically acceptable salt thereof in preparing medicines. Preferably, the medicament is for the prevention or treatment of an EGFR and/or HER2 kinase mediated disease, preferably selected from cancer. Or preferably, the medicament is for the prevention or treatment of cancer and/or the inhibition of metastasis associated with cancer.
According to an embodiment of the invention, the cancer may be selected from a hematological cancer or a solid tumor, such as a solid tumor selected from: lung cancer (e.g., small cell lung cancer or non-small cell lung cancer), papillary thyroid cancer, medullary thyroid cancer, differentiated thyroid cancer, recurrent thyroid cancer, refractory differentiated thyroid cancer, lung adenocarcinoma, bronchiolar lung cancer, multiple endocrine tumors of type 2A or 2B (MEN2A or MEN2B), pheochromocytoma, parathyroid hyperplasia, breast cancer, colorectal cancer (e.g., metastatic colorectal cancer), papillary renal cell carcinoma, ganglioneuroma disease of the gastrointestinal mucosa, inflammatory myofibroblastoma, or cervical cancer. In some embodiments of any of the methods or uses described herein, the cancer is selected from: acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), juvenile cancer, adrenocortical cancer, anal cancer, appendiceal cancer, astrocytoma, atypical teratoma/rhabdoid tumor, basal cell carcinoma, cholangiocarcinoma, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumor, Burkitt's lymphoma, carcinoid tumor, unknown primary cancer, heart tumor, cervical cancer, childhood cancer, chordoma, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), chronic myeloproliferative tumor, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, cholangiocarcinoma, ductal carcinoma in situ, embryonal tumor, endometrial cancer, ependymoma, esophageal cancer, adult neuroblastoma, Ewing sarcoma, extracranial germ cell tumor, extragonadal germ cell tumor, genital cell tumor, adrenal gland tumor, renal cortical tumor, bladder cancer, anal carcinoma, bladder carcinoma, colon cancer, bladder carcinoma, colon carcinoma, bladder carcinoma, Extrahepatic bile duct cancer, ocular cancer, fallopian tube cancer, fibroblastic bone tumor, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor (GIST), germ cell tumor, gestational trophoblastic disease, glioma, hairy cell tumor, hairy cell leukemia, head and neck cancer, cardiac cancer, hepatocellular carcinoma, histiocytosis, hodgkin's lymphoma, hypopharynx cancer, intraocular melanoma, islet cell tumor, pancreatic neuroendocrine tumor, kaposi's sarcoma, kidney cancer, langerhans ' cell histiocytosis, laryngeal cancer, leukemia, lip and oral cancer, liver cancer, lung cancer, lymphoma, macroglobulinemia, malignant fibrous histiocytoma of bone, melanoma, merkel cell cancer, mesothelioma, metastatic neck cancer, midline cancer, squamous cell carcinoma of mouth, multiple endocrine tumor syndrome, multiple myeloma, mycosis fungoides-like granuloma, multiple endocrine tumor, multiple myeloma, mycosis, multiple myeloma, multiple sclerosis, myelodysplastic syndrome, myelodysplastic/myeloproliferative tumors, myelogenous leukemia, multiple myeloma, myeloproliferative tumors, nasal and sinus cancers, nasopharyngeal cancers, neuroblastoma, non-hodgkin's lymphoma, non-small cell lung cancer, oral cancer, lip cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, papillomatosis, paragangliomas, paranasal sinuses and nasal cancers, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pituitary cancer, plasmacytoma, pleuropneumonias, pregnancy and breast cancer, primary central nervous system lymphoma, primary peritoneal cancer, prostate cancer, rectal cancer, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, salivary gland carcinoma, sarcoma, sezary syndrome, skin cancer (such as epidermal carcinoma), small cell lung cancer, small cell carcinoma, cervical cancer, Soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer, gastric cancer, T-cell lymphoma, testicular cancer, throat cancer, thymoma and thymus cancer, thyroid cancer, transitional cell carcinoma of the renal pelvis and ureter, unknown primary cancer, cancer of the urethra, cancer of the uterus, uterine sarcoma, vaginal cancer, cancer of the vulva, and wilms' tumor.
In some embodiments, the patient is a human.
As a medicament, the compounds of the present invention may be administered in the form of a pharmaceutical composition. These compositions may be prepared in a manner well known in the pharmaceutical arts and may be administered by a variety of routes depending on whether local or systemic treatment is desired and the area to be treated. Can be administered topically (e.g., transdermal, dermal, ocular, and mucosal including intranasal, vaginal, and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal), orally, or parenterally. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intracerebroventricular, administration. The administration may be parenteral in a single bolus form, or may be by, for example, a continuous infusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, water, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms (Coated condoms), gloves and the like may also be useful.
In preparing the compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier, for example, in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it may be a solid, semi-solid, or liquid material that serves as a vehicle, carrier, or medium for the active ingredient. Thus, the composition may be in the form of: tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (solid or dissolved in a liquid vehicle); ointments, soft and hard gelatin capsules, suppositories, sterile injectable solutions and sterile packaged powders containing, for example, up to 10% by weight of the active compound.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia gum, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulation may also contain: lubricants such as talc, magnesium stearate and mineral oil; a humectant; emulsifying and suspending agents; preservatives such as methyl benzoate and hydroxypropyl benzoate; sweetening agents and flavoring agents. The compositions of the present invention may be formulated so as to provide immediate, sustained or delayed release of the active ingredient after administration to the patient by employing methods known in the art.
The compositions may be formulated in unit dosage forms, each dosage containing from about 5 to about 1000mg, more usually from about 100 to about 500mg, of the active ingredient. The term "unit dosage form" refers to physically discrete single dosage units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in admixture with suitable pharmaceutical excipients.
The effective dose of the active compound can vary widely and is generally administered in a pharmaceutically effective amount. However, it will be understood that the amount of the compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, and will include the condition to be treated, the chosen route of administration, the actual compound administered; age, weight and response of the individual patient; severity of patient symptoms, etc.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with pharmaceutical excipients to form a solid preformulation composition containing a homogeneous mixture of the compound of the invention. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is generally uniformly distributed throughout the composition such that the composition may be readily divided into equally effective unit dosage forms such as tablets, pills and capsules. The solid pre-formulations are then divided into unit dosage forms of the type described above containing, for example, from about 0.1 to 1000mg of the active ingredient of the invention.
The tablets or pills of the present invention may be coated or compounded to provide a dosage form affording the advantage of prolonged action. For example, a tablet or pill contains an inner dose and an outer dose component, the latter being in the form of a capsule of the former. The two components may be separated by an enteric layer which serves to resist disintegration in the stomach, leaving the inner component intact through the duodenum or delayed in release. A variety of materials may be used for such enteric layers or coatings, such materials including polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
Liquid forms in which the compounds and compositions of the present invention may be incorporated for oral or injectable administration include aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions; and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil; as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions, suspensions, and powders dissolved in pharmaceutically acceptable water or organic solvents or mixtures thereof. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described above. In certain embodiments, the composition is administered by the oral or nasal respiratory route to achieve a local or systemic effect. The composition may be atomized by the use of an inert gas. The nebulized solution may be inhaled directly from the nebulizing device, or the nebulizing device may be connected to a mask or intermittent positive pressure ventilator. The solution, suspension or powder composition may be administered orally or nasally by means of a device that delivers the formulation in a suitable manner.
The amount of compound or composition administered to a patient is not fixed and depends on the drug administered, the purpose of the administration such as prevention or treatment; the condition of the patient, the mode of administration, etc. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. The effective dosage will depend on the disease state being treated and the judgment of the attending clinician, which will depend on factors such as the severity of the disease, the age, weight and general condition of the patient.
Therapeutic dosages of the compounds of the invention may be determined, for example, by: the particular use of the treatment, the mode of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of the compound of the invention in the pharmaceutical composition may not be fixed and will depend on a variety of factors including dosage, chemical properties (e.g., hydrophobicity), and the route of administration. For example, the compounds of the present invention can be provided for parenteral administration by a physiological buffered aqueous solution containing about 0.1-10% w/v of the compound. Some typical dosage ranges are from about 1. mu.g/kg to about 1g/kg body weight/day. In certain embodiments, the dosage range is from about 0.01mg/kg to about 100mg/kg body weight/day. The dosage will likely depend on such variables as the type and extent of progression of the disease or disorder, the general health status of the particular patient, the relative biological efficacy of the selected compound, the excipient formulation and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
Terms and definitions
Unless otherwise indicated, the definitions of groups and terms described in the specification and claims of the present application, including definitions thereof as examples, exemplary definitions, preferred definitions, definitions described in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and coupled with each other. The definitions of the groups and the structures of the compounds in such combinations and after the combination should fall within the scope of the present specification.
Unless otherwise indicated, the numerical ranges set forth in the specification and claims are equivalent to at least each and every specific integer numerical value set forth therein. For example, a numerical range of "1-40" is equivalent to reciting each of the integer values in the numerical range of "1-10," i.e., 1,2, 3,4, 5,6, 7, 8, 9, 10, and each of the integer values in the numerical range of "11-40," i.e., 11, 12, 13, 14, 15, 35, 36, 37, 38, 39, 40. It is to be understood that "more" in one, two, or more of the substituents used herein when describing substituents shall mean an integer ≧ 3, such as 3,4, 5,6, 7, 8, 9, or 10. Further, when certain numerical ranges are defined as "numbers," it should be understood that the two endpoints of the range, each integer within the range, and each decimal within the range are recited. For example, "a number of 0 to 10" should be understood to not only recite each integer of 0, 1,2, 3,4, 5,6, 7, 8, 9, and 10, but also to recite at least the sum of each integer and 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, respectively.
The term "halogen" or "halo" means one of fluorine, chlorine, bromine and iodine.
The term "C1-40Alkyl "is understood to preferably mean a straight-chain or branched saturated monovalent hydrocarbon radical having from 1 to 40 carbon atoms. For example, "C1-6Alkyl "denotes straight-chain and branched alkyl groups having 1,2, 3,4, 5 or 6 carbon atoms. The alkyl group is, for example, a methyl group, an ethyl group, a propyl group, a butyl group, a pentyl group, a hexyl group, an isopropyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an isopentyl group, a 2-methylbutyl group, a 1-ethylpropyl group, a 1, 2-dimethylpropyl group, a neopentyl group, a 1, 1-dimethylpropyl group, a 4-methylpentyl group, a 3-methylpentyl group, a 2-ethylbutyl group, a 1-ethylbutyl group, a 3, 3-dimethylbutyl group, a 2, 2-dimethylbutyl group, a 1, 1-dimethylbutyl group, a 2, 3-dimethylbutyl group, a 1, 3-dimethylbutyl group or a 1, 2-dimethylbutyl group.
The term "C2-40Alkenyl "is understood to preferably mean a straight-chain or branched monovalent hydrocarbon radical comprising one or more double bonds and having from 2 to 40 carbon atoms, preferably" C2-6Alkenyl ". "C2-6Alkenyl "is understood to preferably mean a straight-chain or branched monovalent hydrocarbon radical which contains one or more double bonds and has 2,3, 4,5 or 6 carbon atoms, in particular 2 or 3 carbon atoms (" C)2-3Alkenyl "), it being understood that in the case where the alkenyl group comprises more than one double bond, the double bonds may be separated from each other or conjugated. The alkenyl group is, for example, vinyl, allyl, (E) -2-methylvinyl, (Z) -2-methylvinyl, (E) -but-2-enyl, (Z) -but-2-enyl, (E) -but-1-enyl, (Z) -but-1-enyl, pent-4-enyl, (E) -pent-3-enyl, (Z) -pent-3-enyl, (E) -pent-2-enyl, (Z) -pent-2-enyl, (E) -pent-1-enyl, (Z) -pent-1-enyl, hex-5-enyl, (E) -hex-4-enyl, (Z) -hex-4-enyl, m-n-2-enyl, m-n-1-enyl, m-n-E-4-enyl, m-n-2-, (E) -hex-3-enyl, (Z) -hex-3-enyl, (E) -hex-2-enyl, (Z) -hex-2-enyl, (E) -hex-1-enyl, (Z) -hex-1-enyl, isopropenyl, 2-methylprop-2-enyl, 1-methylprop-2-enyl, 2-methylprop-1-enyl, (E) -1-methylprop-1-enyl, (Z) -1-methylprop-1-enyl, 3-methylbut-3-enyl, 2-methylbut-3-enyl, 1-methylbut-3-enyl, 3-methylbut-2-enyl, (E) -2-methylbut-2-enyl, (Z) -2-methylbut-2-enyl, (E) -1-methylbut-2-enyl, (Z) -1-methylbut-2-enyl, (E) -3-methylbut-1-enyl, (Z) -3-methylbut-1-enyl, (E) -2-methylbut-1-enyl, (Z) -2-methylbut-1-enyl, (E) -1-methylbut-1-enyl, (Z) -1-methylbut-1-enyl, 1-dimethylprop-2-enyl, 1-ethylprop-1-enyl, 1-propylvinyl group and 1-isopropylvinyl group.
The term "C3-40Cycloalkyl is understood to mean a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane having 3 to 40 carbon atoms, preferably "C3-10Cycloalkyl groups ". The term "C3-10Cycloalkyl "is understood to mean a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane having 3,4, 5,6, 7, 8, 9 or 10 carbon atoms. Said C is3-10Cycloalkyl groups may be monocyclic hydrocarbon groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or bicyclic hydrocarbon groups such as decalin rings.
The term "3-20 membered heterocyclyl" means a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane, which contains from 1 to 5 heteroatoms independently selected from N, O and S as a total ring member having from 3 to 20 (e.g., having 3,4, 5,6, 7, 8, 9, 10, etc.) non-aromatic cyclic groups, preferably "3-10 membered heterocyclyl". The term "3-10 membered heterocyclyl" means a saturated monovalent monocyclic, bicyclic hydrocarbon ring or bridged cycloalkane comprising 1-5, preferably 1-3 heteroatoms selected from N, O and S. The heterocyclic group may be attached to the rest of the molecule through any of the carbon atoms or nitrogen atom (if present). In particular, the heterocyclic group may include, but is not limited to: 4-membered rings such as azetidinyl, oxetanyl; 5-membered rings such as tetrahydrofuranyl, dioxolyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl; or a 6-membered ring such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or trithianyl; or a 7-membered ring such as diazepanyl. Optionally, the heterocyclic group may be benzo-fused. The heterocyclyl group may be bicyclic, for example but not limited to a 5,5 membered ring, such as a hexahydrocyclopenta [ c ] pyrrol-2 (1H) -yl ring, or a 5,6 membered bicyclic ring, such as a hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl ring. The nitrogen atom containing ring may be partially unsaturated, i.e., it may contain one or more double bonds, such as, but not limited to, 2, 5-dihydro-1H-pyrrolyl, 4H- [1,3,4] thiadiazinyl, 4, 5-dihydrooxazolyl, or 4H- [1,4] thiazinyl, or it may be benzo-fused, such as, but not limited to, dihydroisoquinolinyl. According to the invention, the heterocyclic radical is non-aromatic. When the 3-20 membered heterocyclic group is linked to another group to form the compound of the present invention, the carbon atom of the 3-20 membered heterocyclic group may be linked to another group, or the heterocyclic atom of the 3-20 membered heterocyclic ring may be linked to another group. For example, when the 3-20 membered heterocyclic group is selected from piperazinyl, it may be such that the nitrogen atom on the piperazinyl group is attached to another group. Or when the 3-20 membered heterocyclyl group is selected from piperidinyl, it may be that the nitrogen atom on the piperidinyl ring and the carbon atom in the para position are attached to other groups.
The term "C6-20Aryl "is understood to preferably mean a monovalent aromatic or partially aromatic radical having from 6 to 20 carbon atomsAromatic monocyclic, bicyclic or tricyclic hydrocarbon rings, preferably "C6-14Aryl ". The term "C6-14Aryl "is to be understood as preferably meaning a mono-, bi-or tricyclic hydrocarbon ring having a monovalent or partially aromatic character with 6, 7, 8, 9, 10, 11, 12, 13 or 14 carbon atoms (" C6-14Aryl group "), in particular a ring having 6 carbon atoms (" C6Aryl "), such as phenyl; or biphenyl, or is a ring having 9 carbon atoms ("C9Aryl), such as indanyl or indenyl, or a ring having 10 carbon atoms ("C10Aryl radicals), such as tetralinyl, dihydronaphthyl or naphthyl, or rings having 13 carbon atoms ("C13Aryl radicals), such as the fluorenyl radical, or a ring having 14 carbon atoms ("C)14Aryl), such as anthracenyl. When said C is6-20When the aryl group is substituted, it may be mono-or polysubstituted. And, the substitution site thereof is not limited, and may be, for example, ortho-, para-or meta-substitution.
The term "5-20 membered heteroaryl" is understood to include such monovalent monocyclic, bicyclic or tricyclic aromatic ring systems: having 5 to 20 ring atoms and comprising 1 to 5 heteroatoms independently selected from N, O and S, such as "5-14 membered heteroaryl". The term "5-14 membered heteroaryl" is understood to include such monovalent monocyclic, bicyclic or tricyclic aromatic ring systems: which has 5,6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, in particular 5 or 6 or 9 or 10 carbon atoms, and which comprises 1 to 5, preferably 1 to 3, heteroatoms each independently selected from N, O and S and, in addition, can be benzo-fused in each case. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, thia-4H-pyrazolyl and the like and their benzo derivatives, such as benzofuryl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl and the like; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof, such as quinolyl, quinazolinyl, isoquinolyl, and the like; or azocinyl, indolizinyl, purinyl and the like and benzo derivatives thereof; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and the like. When the 5-20 membered heteroaryl group is linked to another group to form the compound of the present invention, the carbon atom on the 5-20 membered heteroaryl ring may be linked to another group, or the heteroatom on the 5-20 membered heteroaryl ring may be linked to another group. When the 5-20 membered heteroaryl group is substituted, it may be mono-or poly-substituted. And, there is no limitation on the substitution site thereof, and for example, hydrogen bonded to a carbon atom on a heteroaryl ring may be substituted, or hydrogen bonded to a heteroatom on a heteroaryl ring may be substituted.
Unless otherwise indicated, heterocyclyl, heteroaryl or heteroarylene include all possible isomeric forms thereof, e.g., positional isomers thereof. Thus, for some illustrative non-limiting examples, forms may be included that are substituted at one, two or more of their 1-, 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, 10-, 11-, 12-positions, etc. (if present) or bonded to other groups, including pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, and pyridin-4-yl; thienyl or thienylene includes thien-2-yl, thien-3-yl and thien-3-yl; pyrazol-1-yl, pyrazol-3-yl, pyrazol-4-yl, and pyrazol-5-yl.
The term "oxo" refers to an oxy substitution (═ O) formed by oxidation of a carbon atom, a nitrogen atom or a sulfur atom in a substituent.
Unless otherwise indicated, the definitions of terms herein apply equally to groups comprising the term, e.g. C1-6The definition of alkyl also applies to C1-6Alkyloxy, -N (C)1-6Alkyl radical)2、-NHC1-6Alkyl or-S (O)2-C1-6Alkyl groups, and the like.
It will be appreciated by those skilled in the art that the compounds of formula I may exist in the form of various pharmaceutically acceptable salts. If these compounds have a basic center, they can form acid addition salts; if these compounds have an acidic center, they can form base addition salts; these compounds may also form inner salts if they contain both an acidic centre (e.g. carboxyl) and a basic centre (e.g. amino). The number of salt formation of the compounds in this application is determined by the basic or acidic centers. For example, when a compound contains multiple salt formation sites, the number of salt formation is equal to the number of salt formation sites. By way of example, acid addition salts include, but are not limited to: hydrochloride, hydrofluoride, hydrobromide, hydroiodide, sulfate, pyrosulfate, phosphate, nitrate, methanesulfonate, ethanesulfonate, 2-hydroxyethanesulfonate, benzenesulfonate, toluenesulfonate, sulfamate, 2-naphthalenesulfonate, formate, acetoacetic acid, pyruvic acid, lauric acid ester, cinnamate, benzoate, acetate, glyoxylate, trifluoroacetate, pivalate, propionate, butyrate, hexanoate, heptanoate, undecanoate, stearate, ascorbate, camphorate, camphorsulfonate, citrate, fumarate, malate, maleate, hydroxymaleate, oxalate, salicylate, succinate, gluconate, quinic acid, pamoate, glycolate, tartrate, lactate, 2- (4-hydroxybenzoyl) benzoate, Cyclopentane propionate, digluconate, 3-hydroxy-2-naphthoate, nicotinate, embonate, pectinate, 3-phenylpropionate, picrate, pivalate, itaconate, triflate, lauryl sulfate, p-toluenesulfonate, napadisylate, malonate, adipate, alginate, mandelate, glucoheptonate, glycerophosphate, sulfosalicylate, hemisulfate or thiocyanate, aspartate, and the like.
Depending on their molecular structure, the compounds of the invention exist as stereoisomers, which may be, for example, chiral or cis-trans isomeric, and may thus exist in the form of various enantiomers or diastereomers.
For cis-trans isomers, the two atoms or groups attached to each carbon atom should be compared according to the "order rules" known to those skilled in the art, with the two preferred groups being Z isomers on the same side of the pi bond plane and E isomers on the opposite side.
For chiral isomers, they may exist in racemic or optically active form. The compounds of the invention or intermediates thereof may be separated into enantiomeric compounds by chemical or physical methods well known to those skilled in the art, or used in this form for synthesis. In the case of racemic amines, diastereomers are prepared from mixtures by reaction with optically active resolving agents. Examples of suitable resolving agents are optically active acids such as the R and S forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid, suitable N-protected amino acids (e.g. N-benzoylproline or N-benzenesulfonylproline) or various optically active camphorsulphonic acids. The chromatographic enantiomeric resolution can also advantageously be carried out with the aid of optically active resolving agents, such as dinitrobenzoylphenylglycine, cellulose triacetate or other carbohydrate derivatives or chirally derivatized methacrylate polymers, which are immobilized on silica gel. Suitable eluents for this purpose are aqueous or alcoholic solvent mixtures, for example hexane/isopropanol/acetonitrile. The corresponding stable isomers can be isolated according to known methods, for example by extraction, filtration or column chromatography.
The term "patient" refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses or primates, most preferably humans.
The phrase "therapeutically effective amount" as used herein refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that a researcher, veterinarian, medical doctor or other clinician is seeking in a tissue, system, animal, individual, or human, which includes one or more of the following: (1) prevention of diseases: for example, preventing a disease, disorder or condition in an individual who is susceptible to the disease, disorder or condition but has not experienced or developed disease pathology or symptomatology. (2) Inhibiting the disease: for example, inhibiting the disease, disorder or condition (i.e., arresting the further development of the pathology and/or condition) in an individual who is experiencing or presenting the pathology or condition of the disease, disorder or condition. (3) And (3) relieving the diseases: for example, relieving the disease, disorder or condition (i.e., reversing the pathology and/or symptomatology) in an individual who is experiencing or presenting with the pathology or symptomatology of the disease, disorder or condition.
Advantageous effects
The inventor surprisingly finds that, different from various existing quinoline inhibitors, the compound of the invention adopts a nitrogen-containing heterocyclic group (especially pyridyl) with wide biological activity at the 6-position of the quinoline skeleton for substitution, and simultaneously modifies the substituent at the 4-position of the quinoline skeleton, so as to design and synthesize a novel anti-tumor active compound. Compared with the existing antitumor drugs, the compounds have excellent antitumor activity on A431, SKOV3, SK-BR-3, BT474 and the like, and have good application prospects. In addition, the preparation method of the compound has the advantages of low cost, simple steps, mild reaction conditions, high yield and easy post-treatment.
Detailed Description
The technical solution of the present invention will be further described in detail with reference to specific embodiments. It is to be understood that the following examples are only illustrative and explanatory of the present invention and should not be construed as limiting the scope of the present invention. All the technologies realized based on the above-mentioned contents of the present invention are covered in the protection scope of the present invention.
Unless otherwise indicated, the raw materials and reagents used in the following examples are all commercially available products or can be prepared by known methods.
Example 1: (E) synthesis of (a) -N- (4- (4- (benzyloxy) anilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide
Figure BDA0002878186340000201
(1) Synthesis of trans-3- (3-pyridyl) acryloyl chloride (a-1)
A50 mL three-necked flask equipped with a thermometer and equipped with an electromagnetic stirrer was charged with trans-3- (3-pyridyl) acrylic acid (0.003mol,0.45g) and 7mL of thionyl chloride, and the mixture was refluxed for 4 hours under nitrogen protection, and the remaining solvent was distilled off under reduced pressure to remove the solvent under reduced pressure, whereby 0.46g of a pale yellow oil was obtained in a yield of 91.0%.
(2) Synthesis of N- (4- (4- (benzyloxy) phenylamino) -3-cyano-7-ethoxyquinolin-6-yl) acetamide (a-2)
To a 500mL three necked flask equipped with a thermometer, magnetic stirrer was added N- (4-chloro-3-cyano-7-ethoxyquinolin-6-yl) acetamide (0.024mol,6.90g),4- (benzyloxy) aniline (0.026mol,5.17g) and (0.026mol,3.0g) pyridine hydrochloride, 250mL isopropanol, as a developing solvent petroleum ether: ethyl acetate volume ratio 1: stirring, refluxing for 12h, standing, filtering, and recrystallizing with methanol to obtain 9.05g and 83% of N- (4- (4- (benzyloxy) phenylamino) -3-cyano-7-ethoxyquinolin-6-yl) acetamide.
1H NMR(DMSO-d6):δ11.01(brs,1H),9.61(s,1H),9.05(s,1H),8.94(s,1H),7.49-7.50(m,3H),7.33-7.474(m,5H),7.12(d,J=8.8Hz,2H),5.16(s,2H)4.33(q,J=6.8Hz,2H),2.19(s,3H)1.49(t,J=6.8Hz,3H)。
(3) Synthesis of 4- (4- (benzyloxy) aniline) -6-amino-7-ethoxyquinoline-3-carbonitrile (a-3) N- (4- (4- (benzyloxy) phenylamino) -3-cyano-7-ethoxyquinolin-6-yl) acetamide (0.023mol,10.0g) was dissolved in 200mL methanol, HCl (40mL 2N) was added, 60 ℃, TLC follow-up, and the developing solvent was petroleum ether: ethyl acetate volume ratio 1: 2, reacting for 24 hours, removing residual solvent by rotary evaporation, stirring residual solid for 1 hour by using saturated sodium bicarbonate aqueous solution, filtering, and recrystallizing a filter cake by using methanol to obtain 8.95g of yellow solid with the yield of 90 percent.
1H NMR(DMSO-d6):δ10.62(S,1H),8.77(s,1H),7.57(s,1H),7.48(d,J=7.2Hz,2H),7.33-7.43(m,6H),7.08-7.11(m,2H),5.48(br,S,2H)5.16(s,2H),4.26(q,J=6.8Hz,2H),1.48(t,J=6.8Hz,3H)
(4) Synthesis of (E) -N- (4- (4- (benzyloxy) anilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide Synthesis of (a-4)
Trans-3- (3-pyridyl) acryloyl chloride (a-1) (0.43g, 0.0026mol) was dissolved in N-methylpyrrolidone (3mL) and transferred to a constant pressure titration funnel, a solution of 4- (4- (benzyloxy) aniline) -6-amino-7-ethoxyquinoline-3-carbonitrile (0.002mol,0.82g) in N-methylpyrrolidone (2mL) was added dropwise, the reaction was carried out at 0 ℃ under nitrogen, followed by TLC with a developing solvent of petroleum ether: ethyl acetate volume ratio 1:1 for 2 hours, stirring the reaction solution by using a saturated sodium bicarbonate solution to adjust the pH value to 11, performing suction filtration, and mixing a filter cake by using dichloromethane and methanol in a volume ratio of 1: 4 to give (E) -N- (4- (4- (benzyloxy) anilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide (0.99g, 91%).
1H NMR(DMSO-d6):δ9.60(s,2H),9.05(s,1H),8.85(d,J=1.6Hz,1H),8.58-8.60(dd,J=5.6,1.6Hz,1H),8.43(s,1H),8.08(d,J=8Hz,1H),7.64(d,J=16Hz,1H),7.32-7.51(m,8H),7.22(d,J=8.8Hz,2H),7.05(d,J=5.2Hz,2H),5.12(s,2H),4.35(q,J=6.8Hz,2H),1.50(t,J=6.8Hz,3H)
13C NMR(DMSO-d6):δ163.42,156.36,153.05,152.64,150.94,150.47,149.36,147.59,137.23,136.99,134.22,132.87,130.52,128.39,127.80,127.72,127.40,126.24,124.25,124.00,117.22,115.14,115.11,115.06,112.96,108.76,86.47,69.46,64.57,14.27
ESI-HRMS cacld.For C33H27N5O3[M+H]+:542.2192,found 542.2185
Example 2: (E) synthesis of (b) -N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide
Figure BDA0002878186340000221
(1) Synthesis of N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) acetamide (b-1)
According to the method of step (2) in example 1, N- (4-chloro-3-cyano-7-ethoxyquinolin-6-yl) acetamide (0.024mol,6.90g), 4-chloroaniline (0.026mol,3.30g), and (0.026mol, 3.00 g) pyridine hydrochloride, 250mL of isopropanol were charged into a 500mL three-necked flask equipped with a thermometer and electromagnetic stirrer, followed by TLC detection using petroleum ether as a developing solvent: ethyl acetate volume ratio 1:1, stirring, refluxing, reacting for 12h, standing, filtering, and recrystallizing with methanol to obtain intermediate b-1(7.90g, 87%).
1H NMR(DMSO-d6):δ11.09(br,s,1H),9.62(s,1H),9.08(s,1H),9.01(s,1H),7.62(s,1H),7.53-7.56(m,2H),7.44-7.47(m,2H),4.33(q,J=6.8Hz,2H),2.11(s,3H),1.50(t,J=6.8Hz,3H)
(2) Synthesis of 4- (4-chloroanilino) -6-amino-7-ethoxyquinoline-3-carbonitrile (b-2)
According to the method of step (3) in example 1, (0.023mol,8.74g) N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) acetamide (b-1) was dissolved in 200mL methanol, HCl (40mL 2N) was added, 60 ℃, TLC follow-up, and the developing solvent was petroleum ether: ethyl acetate volume ratio 1: 2, reacting for 24h, removing residual solvent by rotary evaporation, stirring residual solid for 1h by using saturated sodium bicarbonate aqueous solution, filtering, and recrystallizing a filter cake by using methanol to obtain 7.15g of yellow solid with the yield of 92%.
1H NMR(DMSO-d6):δ10.72(s,1H),8.83(s,1H),7.41-7.55(m,6H),5.95(br,s,2H),4.26(q,J=6.8Hz,2H),1.49(t,J=6.8Hz,3H)
(3) (b-3) Synthesis of (E) -N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide
According to the method of step (4) in example 1, trans-3- (3-pyridyl) acryloyl chloride (a-1) (0.43g, 0.0026mol) was dissolved in N-methylpyrrolidone (3mL) and transferred to a constant pressure titration funnel, and a solution of 4- (4-chloroaniline) -6-amino-7-ethoxyquinoline-3-carbonitrile (0.68g,0.002mol) in N-methylpyrrolidone (2mL) was added dropwise, with nitrogen protection at 0 ℃, TLC tracing, and the developing solvent was petroleum ether: ethyl acetate volume ratio 1:1 for 2 hours, stirring the reaction solution by using a saturated sodium bicarbonate solution to adjust the pH value to 11, performing suction filtration, and mixing a filter cake by using dichloromethane and methanol in a volume ratio of 1: 4 to give (E) -N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-2-yl) acrylamide (0.86g, 92%).1H NMR(DMSO-d6):δ9.69(s,1H),9.57(s,1H),9.09(s,1H),8.85(s,1H),8.58-8.60(m,2H),8.07(d,J=8.0Hz,1H),7.64(d,J=15.6Hz,1H),7.21-7.50(m,7H),4.36(q,J=6.8Hz,2H),1.50(t,J=6.8Hz,3H)
13C NMR(DMSO-d6):δ163.55,153.00,151.92,150.48,149.82,149.37,147.83,139.80,137.44,134.26,130.51,128.84,128.08,127.70,124.20,123.99,123.48,117.05,114.36,108.65,89.88,64.75,14.24
ESI-HRMS cacld.For C26H20ClN5O2[M+H]+:470.1384,found 470.1377
Example 3: (E) synthesis of (E) -N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide (c)
Figure BDA0002878186340000241
(1) Synthesis of N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) acetamide (c-1)
According to the method of step (2) in example 1, N- (4-chloro-3-cyano-7-ethoxyquinolin-6-yl) acetamide (0.024mol,6.90g),4- (3-fluorobenzyloxy) -3-chloroaniline (0.026mol,6.53g) and (0.026mol,3.0g) pyridine hydrochloride, 250mL of isopropanol were charged in a 500mL three-necked flask equipped with a thermometer and electromagnetic stirrer, and the developing solvent was petroleum ether: ethyl acetate volume ratio 1:1, stirred and refluxed for 12h, left to stand, filtered with suction, and recrystallized from methanol to give intermediate c-1(10.52g, 87%).
1H NMR(DMSO-d6):δ11.09(br,s,1H),9.62(s,1H),8.98-9.10(m,2H)7.59-7.63(m,2H)7.27-7.50(m,5H),7.17-7.21(m,1H),5.31(s,2H)4.33(q,J=6.8Hz,2H),2.19(s,3H)1.50(t,J=6.8Hz,3H)
(2) Synthesis of 4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -6-amino-7-ethoxyquinoline-3-carbonitrile (c-2)
According to the procedure of step (3) in example 1, (0.023mol,11.59g) N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) acetamide (c-1) was dissolved in 200mL of methanol, HCl (40mL of 2N) was added, 60 ℃ was monitored by TLC tracing and the developing solvent was petroleum ether: ethyl acetate volume ratio 1: 2, reacting for 24 hours, removing residual solvent by rotary evaporation, stirring the residual solid for 1 hour by using saturated sodium bicarbonate aqueous solution, filtering, and recrystallizing a filter cake by using methanol to obtain 9.82g of yellow solid with the yield of 93 percent.
1H NMR(DMSO-d6):δ10.70(s,1H),8.79(s,1H),7.29-7.50(m,8H),7.17-7.21(m,1H),5.94(b,2H)5.30(s,2H),4.26(q,J=6.8Hz,2H),1.48(t,J=6.8Hz,3H)
(3) (c-3) Synthesis of (E) -N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide
According to the method of step (4) in example 1, trans-3- (3-pyridyl) acryloyl chloride (a-1) (0.43g, 0.0026mol) was dissolved in N-methylpyrrolidone (3mL) and transferred to a constant pressure titration funnel, and a solution of 4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -6-amino-7-ethoxyquinoline-3-carbonitrile (0.92g,0.002mol) in N-methylpyrrolidone (2mL) was added dropwise, under nitrogen protection at 0 ℃, followed by TLC, and the developing solvent was petroleum ether: ethyl acetate volume ratio 1:1 for 2 hours, stirring the reaction solution by using a saturated sodium bicarbonate solution to adjust the pH value to 11, filtering, and mixing a filter cake by using dichloromethane and methanol in a volume ratio of 1: the mixture of 4 was recrystallized to give 1.06g (90%) of (E) -N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-2-yl) acrylamide.
1H NMR(DMSO-d6):δ9.67(s,1H),9.61(s,1H),9.06(s,1H),8.85(d,J=1.6Hz,1H),8.60(dd,J=4.8,1.6Hz,1H),8.49(s,1H),8.07(d,J=8.0Hz,1H),7.64(d,J=15.6Hz,1H),7.30-7.66(m,7H),7.16-7.26(m,3H),5.26(s,2H),4.36(q,J=6.8Hz,2H),1.50(t,J=6.8Hz,3H)
13C NMR(DMSO-d6):δ163.47,163.38,160.96,153.01,152.36,150.99,150.48,149.37,147.66,139.49,137.32,134.23,134.03,130.51,127.71,125.75,124.24,124.00,123.34,117.20,114.77,114.56,114.44,114.14,113.92,113.30,108.75,87.48,69.43,64.65,14.26
ESI-HRMS cacld.For C33H25ClFN5O3[M+H]+:594.1708,found 594.1702
In vitro antitumor Activity test example
The principle is as follows: succinate dehydrogenase in mitochondria of living cells can reduce thiazole blue (MTT) to water-insoluble blue-purple needle-shaped crystalline Formazan (Formazan) and deposit in cells, while dead cells do not. Dimethyl sulfoxide (DMSO) dissolves bluish-purple crystals in cells and has a maximum absorption peak at 490 nm. Thus, the number of living cells can be reflected by measuring the absorbance at 490nm wavelength. The amount of formazan formation is directly proportional to the number of cells over a range of cell numbers.
Test cell
Human epidermal carcinoma cell A431, breast cancer cells SKBR3 and BT474, and ovarian cancer cell strain SKOV3, wherein the A431 cell is purchased from Shanghai academy of sciences cell bank, the SK-BR-3 cell is purchased from Wuhan Punuoise Life technologies, Inc., SKOV3, and the BT474 cell is purchased from Shanghai Saibei sweetgum biotechnology, Inc.
Main instrument and consumable
Biosafety cabinets, model HR60-II42, Haier, China
CO2Incubator, model IL-161HT, Shanghai, China
Inverted biomicroscope model CKX41SF, Olympus, Japan
Air bath constant temperature shaking table, Guohua corporation, model ZD-85
Full-automatic multifunctional microplate reader manufactured by Bio-Rad of America
Precision adjustable micropipettes, Eppendorf (germany);
96-well cell culture plates, Corning (usa);
cell culture flasks, Corning (USA).
Primary reagent
Fetal bovine serum, GEMINI corporation;
trypsin, Gibco;
RPMI1640 medium (Beijing Solarbio Science & Technology Co., Ltd.), DMEM medium (Beijing Solarbio Science & Technology Co., Ltd.), DMSO and tetramethylazotolium blue (MTT) Sigma USA.
The experimental procedure was as follows:
cell culture
DMEM culture medium for human epidermal cancer cell A431 and breast cancer SKBR3 cell, 1640 culture medium for ovarian cancer cell line SKOV3 and breast cancer BT474 cell, and saturated humidity at 37 deg.C and 5% CO2Performing conventional culture in an incubator; the medium was changed 1 time every 2-3d, and when the cells grew to logarithmic phase, subculture was performed. Cell activity was detected during the experiment using trypan blue staining.
Preparing working solution
Preparation of MTT solution: MTT50mg was weighed, dissolved in 10mL Phosphate Buffered Saline (PBS), sterilized by filtration through a 0.22 μm millipore filter, dispensed, stored at 4 ℃ in the dark and effective for two weeks.
Preparation of test drugs: the compounds a-c prepared in examples 1-3 and DMSO were dissolved in 50mmol/L of stock solution, and the stock solution was diluted with culture medium to different concentrations of working solution, such as 50. mu. mol/L, 25. mu. mol/L, 12.5. mu. mol/L, 6.25. mu. mol/L, and 3.125. mu. mol/L, by precise aspiration before use.
Measurement method
1. Taking logarithmic phase A431, SKBR3, SKOV3 and BT474 cells, digesting, centrifuging and resuspending, respectively adjusting the cell density to 8 x 1041X 10 units/mL52X 10 units/mL5one/mL was seeded in 96-well cell culture plates, where 100. mu.l of the prepared cell suspension was added per well and then placed in a cell incubator for culture.
2. After waiting overnight, i.e., after the cells were adherent, the upper layer of culture was discarded. The test group was added with 100. mu.l of compound (a-c) at concentrations of 50. mu. mol/L, 25. mu. mol/L, 12.5. mu. mol/L, 6.25. mu. mol/L, and 3.125. mu. mol/L, respectively, 3 duplicate wells per concentration, and a negative control group (to which an equal volume of cell suspension was added) and a blank control group (to which an equal volume of medium was added).
3. After culturing the 96-well plates for 48 hours, 20. mu.l of MTT solution (5 mg/ml) was added to each well, and the culture was continued in the incubator for 4 hours.
4. The 96-well plate was taken out of the incubator, the medium in the well was carefully aspirated off by a micropipette, 150. mu.L of dimethyl sulfoxide was added to each well, the crystals were completely dissolved by shaking for 10 minutes on a horizontal shaker, and then the absorbance OD value was measured at a wavelength of 490nm using a microplate reader.
5. The test was repeated three times in succession, and the cell inhibition rate was calculated according to the following formula: cell inhibition rate (%) [1- (test OD value-blank OD)/(negative control OD value-blank OD)]X 100%, and calculating the half inhibitory concentration IC of the sample for inhibiting cell growth50The value is obtained.
The results of the experiments showed that less than 0.1 v% DMSO solvent had negligible effect on the cells, IC50 values were calculated using the statistical software spss13.0, and the results are summarized in table 1.
Table 1: in vitro antitumor Activity of target Compounds IC50Data (. mu. mol/L)
Figure BDA0002878186340000281
Figure BDA0002878186340000291
The results show that the compounds of the embodiment of the invention have significant inhibitory activity on tumor cells A431, SKBR3, SKOV3 and BT 474. Among them, the compounds a and c have more excellent inhibitory activity against tumor cells SKBR3 and BT 474.

Claims (10)

1. A compound of formula I, a stereoisomer, or a pharmaceutically acceptable salt thereof:
Figure FDA0002878186330000011
wherein R is1Selected from CN, halogen, OH, SH, unsubstituted or substituted by one or more RaSubstituted of the following groups: c1-40Alkyl radical, C1-40Alkyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each Y is the same or different and is independently selected from a bond or-O-;
R2、R3identical or different, independently of one another, from H, unsubstituted or substituted by one or more RbSubstituted of the following groups: c1-40Alkyl radical, C3-40Cycloalkyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl;
R4selected from unsubstituted or substituted by one or more RcSubstituted of the following groups: a 5-20 membered heteroaryl or 3-20 membered heterocyclyl, provided that at least 1 ring atom of the heteroaryl or heterocyclyl is N;
R5selected from unsubstituted or substituted by one or more RdSubstituted C1-40An alkyl group;
each Ra、Rb、Rc、RdIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2Unsubstituted or substituted by one or more ReSubstituted of the following groups: c1-40Alkyl radical, C2-40Alkenyl radical, C3-40Cycloalkyl radical, C3-40Cycloalkenyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, C1-40Alkyloxy, C2-40Alkenyloxy radical, C3-40Cycloalkyl oxy, C3-40Cycloalkenyloxy, C6-20Aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each ReIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2Unsubstituted or substituted by one or more RfSubstituted of the following groups: c1-40Alkyl radical, C2-40Alkenyl radical, C3-40Cycloalkyl radical, C3-40Cycloalkenyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, C1-40Alkyloxy, C2-40Alkenyloxy radical, C3-40Cycloalkyl oxy, C3-40Cycloalkenyloxy, C6-20Aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each RfIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2、C1-40Alkyl radical, C2-40Alkenyl radical, C3-40Cycloalkyl radical, C3-40Cycloalkenyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl, C1-40Alkyloxy, C2-40Alkenyloxy radical, C3-40Cycloalkyl oxy, C3-40Cycloalkenyloxy, C6-20Aryloxy, 5-20 membered heteroaryloxy, 3-20 membered heterocyclyloxy, NH2、-Y-C(O)R6、-C(O)-Y-R6、-Y-S(O)2R7、-S(O)2-Y-R7
Each R6、R7Same or different, independently from each other selected from H, C1-40Alkyl radical, C3-40Cycloalkyl radical, C6-20Aryl, 5-20 membered heteroaryl, 3-20 membered heterocyclyl.
2. The compound of claim 1, a stereoisomer, or pharmaceutically acceptable salt thereof, wherein the compound of formula I has a structure as shown in formula I':
Figure FDA0002878186330000021
wherein R is1、R2、R3、R4、R5Having the definition set forth in claim 1.
3. The compound of claim 1 or 2, a stereoisomer, or pharmaceutically acceptable salt thereof, wherein:
R1selected from CN, halogen, OH, SH, C1-6Alkyl, NH2、COOH、-OC(O)C1-6Alkyl, -C(O)OC1-6An alkyl group;
R2、R3identical or different, independently of one another, from H or by one or more RbSubstituted C6-10An aryl group;
Rbselected from halogen, CN, OH, SH, NO2Unsubstituted or substituted by one or more ReSubstituted of the following groups: c1-6Alkyl radical, C1-6An alkyloxy group;
Reselected from halogen, CN, OH, SH, NO2Unsubstituted or substituted by one or more RfSubstituted C6-10An aryl group;
Rfselected from halogens.
4. A compound, stereoisomer or pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-3, wherein:
R2selected from 4- (benzyloxy) phenyl, 4-chlorophenyl, 4- (3-fluorobenzyloxy) -3-chlorophenyl;
R4selected from unsubstituted or substituted by one or more RcA substituted 5-6 membered heteroaryl or 5-6 membered heterocyclyl, provided that 1 or 2 ring atoms of the heteroaryl or heterocyclyl are N; for example, R4Selected from unsubstituted or substituted by one or more RcSubstituted of the following groups: pyridin-1-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl;
R5is selected from C1-6An alkyl group;
each Ra、Rb、Rc、RdIdentical or different, independently of one another, from halogen, CN, OH, SH, oxo (═ O), NO2、C1-6Alkyl radical, C1-6Alkyloxy, C6-10Aryloxy, 5-6 membered heteroaryloxy, 5-6 membered heterocyclyloxy.
5. The compound of any one of claims 1-4, a stereoisomer, or pharmaceutically acceptable salt thereof, wherein the compound of formula I has a structure as shown in formula I-1 below:
Figure FDA0002878186330000041
alternatively, the compounds of formula I have the structure shown in formula I-2 below:
Figure FDA0002878186330000042
wherein R is1、R2、R3、R5Having the definition as set forth in any one of claims 1 to 4.
6. The compound of claim 1, a stereoisomer, or pharmaceutically acceptable salt thereof, wherein the compound of formula I is selected from the group consisting of:
(E) -N- (4- (4- (benzyloxy) anilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide;
(E) -N- (4- (4-chloroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide;
(E) -N- (4- (4- (3-fluorobenzyloxy) -3-chloroaniline) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide;
(E) -N- (4- (4-fluoroanilino) -3-cyano-7-ethoxyquinolin-6-yl) -3- (pyridin-3-yl) acrylamide.
7. A process for the preparation of a compound according to any one of claims 1 to 6, a stereoisomer or a pharmaceutically acceptable salt thereof, which comprises reacting a compound of formula II with a compound of formula III to give a compound of formula I:
Figure FDA0002878186330000051
wherein R is1、R2、R3、R4And R5Having the definition of any one of claims 1 to 6;
preferably, the reaction of the compound of formula II with the compound of formula III is carried out in the presence of an organic solvent, which may be selected from N-methylpyrrolidone.
Preferably, the reaction temperature of the compound of formula II with the compound of formula III may be 0-10 ℃; the product of the reaction may be purified by recrystallization, which may be a mixed solvent of dichloromethane and methanol, wherein the volume ratio of dichloromethane to methanol may be 1:1 to 1: 5.
8. The production method according to claim 7, wherein:
the compound of formula II is prepared by reacting a compound of formula IV below with a halogenating agent:
Figure FDA0002878186330000052
wherein R is4Having the definition of any one of claims 1 to 6;
and/or the presence of a gas in the gas,
the compound of formula III is prepared by the reaction of a compound of formula V below with a compound of formula VI below:
Figure FDA0002878186330000053
wherein L is selected from a leaving group, such as Cl, Br or I;
R1、R2、R3、R4and R5Having the definition as set forth in any one of claims 1 to 6.
9. A pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I according to any one of claims 1 to 6, a stereoisomer or a pharmaceutically acceptable salt thereof.
10. Use of a compound of formula I, a stereoisomer or pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 6, in the manufacture of a medicament for the prevention or treatment of an EGFR and/or HER2 kinase mediated disease, or for the prevention or treatment of cancer and/or for inhibiting metastasis associated with cancer;
preferably, the cancer is selected from hematologic cancers or solid tumors, preferably from breast cancer, ovarian cancer and skin cancer.
CN202011640042.3A 2020-12-31 2020-12-31 Pyridyl substituted quinoline derivative and its prepn and use Active CN112625025B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011640042.3A CN112625025B (en) 2020-12-31 2020-12-31 Pyridyl substituted quinoline derivative and its prepn and use

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011640042.3A CN112625025B (en) 2020-12-31 2020-12-31 Pyridyl substituted quinoline derivative and its prepn and use

Publications (2)

Publication Number Publication Date
CN112625025A true CN112625025A (en) 2021-04-09
CN112625025B CN112625025B (en) 2022-03-29

Family

ID=75290170

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011640042.3A Active CN112625025B (en) 2020-12-31 2020-12-31 Pyridyl substituted quinoline derivative and its prepn and use

Country Status (1)

Country Link
CN (1) CN112625025B (en)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101824029A (en) * 2009-03-05 2010-09-08 厦门艾德生物医药科技有限公司 Tyrosine kinase irreversible inhibitor and medicine composition and application thereof
WO2011029265A1 (en) * 2009-09-14 2011-03-17 江苏恒瑞医药股份有限公司 6-amino quinazoline or 3-cyano quinoline derivatives, preparation methods and pharmaceutical uses thereof
WO2011077095A1 (en) * 2009-12-22 2011-06-30 Imperial Innovations Limited Quinoline derivatives used as pet imaging agents
CN102838539A (en) * 2011-06-21 2012-12-26 苏州迈泰生物技术有限公司 Quinolinylenamide derivative and its application in preparation of anti-malignant tumor drugs
WO2013131424A1 (en) * 2012-03-09 2013-09-12 上海恒瑞医药有限公司 4-quinazoline amine derivative and application thereof
WO2017186140A1 (en) * 2016-04-28 2017-11-02 江苏恒瑞医药股份有限公司 Method for preparing tyrosine kinase inhibitor and derivative thereof
CN109422755A (en) * 2017-09-01 2019-03-05 上海医药集团股份有限公司 A kind of nitrogen-containing heterocycle compound, preparation method, intermediate, composition and application
CN112110899A (en) * 2020-10-29 2020-12-22 苏州富士莱医药股份有限公司 Synthetic method of pyrroltinib

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101824029A (en) * 2009-03-05 2010-09-08 厦门艾德生物医药科技有限公司 Tyrosine kinase irreversible inhibitor and medicine composition and application thereof
WO2011029265A1 (en) * 2009-09-14 2011-03-17 江苏恒瑞医药股份有限公司 6-amino quinazoline or 3-cyano quinoline derivatives, preparation methods and pharmaceutical uses thereof
WO2011077095A1 (en) * 2009-12-22 2011-06-30 Imperial Innovations Limited Quinoline derivatives used as pet imaging agents
CN102838539A (en) * 2011-06-21 2012-12-26 苏州迈泰生物技术有限公司 Quinolinylenamide derivative and its application in preparation of anti-malignant tumor drugs
WO2013131424A1 (en) * 2012-03-09 2013-09-12 上海恒瑞医药有限公司 4-quinazoline amine derivative and application thereof
WO2017186140A1 (en) * 2016-04-28 2017-11-02 江苏恒瑞医药股份有限公司 Method for preparing tyrosine kinase inhibitor and derivative thereof
CN109422755A (en) * 2017-09-01 2019-03-05 上海医药集团股份有限公司 A kind of nitrogen-containing heterocycle compound, preparation method, intermediate, composition and application
CN112110899A (en) * 2020-10-29 2020-12-22 苏州富士莱医药股份有限公司 Synthetic method of pyrroltinib

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HWEI-RU TSOU 等: "Optimization of 6,7-Disubstituted-4-(arylamino)quinoline-3-carbonitriles as Orally Active, Irreversible Inhibitors of Human Epidermal Growth Factor Receptor-2 Kinase Activity", 《JOURNAL OF MEDICINAL CHEMISTRY》 *
XIN LI 等: "Discovery and development of pyrotinib: A novel irreversible EGFR/HER2 dual tyrosine kinase inhibitor with favorable safety profiles for the treatment of breast cancer", 《EUROPEAN JOURNAL OF PHARMACEUTICAL SCIENCES》 *

Also Published As

Publication number Publication date
CN112625025B (en) 2022-03-29

Similar Documents

Publication Publication Date Title
RU2682245C1 (en) Indazolie wnt signal pathway inhibitors and their therapeutic applications
CN108026101B (en) Fused pyrimidine compound or salt thereof
JP6355648B2 (en) 3- (Benzimidazol-2-yl) -indazole inhibitors of WNT signaling pathway and their therapeutic use
CA2905993C (en) Substituted 4-amino-pyrimidinyl-2-amino-phenyl derivatives and pharmaceutical compositions thereof for use as jak2 and alk2 inhibitors
TW201930292A (en) Pyridine, pyrazine, and triazine compounds as allosteric SHP2 inhibitors
JP6035423B2 (en) Novel condensed pyrimidine compound or salt thereof
JP6050241B2 (en) Novel compounds for the treatment of neurodegenerative diseases
CN110382499A (en) FGFR inhibitor and its application
WO2016169421A1 (en) Imidazo isoindole derivative, preparation method therefor and medical use thereof
JP2021501215A (en) Amino-substituted nitrogen-containing condensed ring compound, its preparation method and use
WO2020156283A1 (en) Alkynylpyrimidine or alkynylpyridine compound, and composition and application thereof
WO2020020385A1 (en) Tricyclic derivative inhibitor, preparation method therefor, and application
JP2019510055A (en) Compounds for the inhibition of cyclophilin and their use
WO2022017408A1 (en) Arylamine derivative and preparation method therefor and medical use thereof
CN113402543B (en) Butylphthalide ring-opening derivative and preparation method and application thereof
CN111153891B (en) Substituted benzimidazole PI3K alpha/mTOR double-target inhibitor and pharmaceutical composition and application thereof
CN112625025B (en) Pyridyl substituted quinoline derivative and its prepn and use
CN112694439B (en) Phenyl acrylamide quinoline derivative and preparation method and application thereof
CN112759583B (en) Quinoline derivative containing furyl and preparation method and application thereof
CN114057823B (en) Betulinic acid derivative, preparation method, pharmaceutical composition and application thereof
CN114392262A (en) Betulinic acid derivatives for inhibiting nervous system degenerative diseases
CN115894600A (en) Betulol derivative, and preparation method, pharmaceutical composition and application thereof
TWI820414B (en) Quinazoline compounds, preparation method and use thereof
WO2022194265A1 (en) Quinazoline-based compound, composition, and application of quinazoline-based compound
WO2022022630A1 (en) Oxa-azaspiro derivative, and preparation method therefor and pharmaceutical use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant